scholarly journals The miR-26 family regulates neural differentiation-associated microRNAs and mRNAs by directly targeting REST

2021 ◽  
Author(s):  
Mark Sauer ◽  
Nina Was ◽  
Thomas Ziegenhals ◽  
Xiantao Wang ◽  
Markus Hafner ◽  
...  

The repressor element silencing transcription factor (REST) plays a crucial role in the differentiation of neural progenitor cells (NPCs). Effector proteins of REST are C-terminal domain small phosphatases (CTDSPs), which reduce polymerase II activity on genes required for neurogenesis. miR-26b regulates neurogenesis in zebrafish by targeting ctdsp2 mRNA, but the molecular events triggered by this microRNA remain unknown. Here we show in a murine embryonic stem cell differentiation paradigm that inactivation of miR-26 family members disrupts the formation of neurons and astroglia and arrests neurogenesis at the neural progenitor level. We further show that miR-26 directly targets Rest, thereby inducing the expression of a large set of REST complex-repressed neuronal genes including miRs required for the induction of the neuronal gene expression program. Our data identify the miR-26 family as the trigger of a self-amplifying system required for neural differentiation that acts upstream of REST-controlled miRs.

2007 ◽  
Vol 6 (11) ◽  
pp. 4374-4387 ◽  
Author(s):  
Alison V. Nairn ◽  
Akiko Kinoshita-Toyoda ◽  
Hidenao Toyoda ◽  
Jin Xie ◽  
Kyle Harris ◽  
...  

2011 ◽  
Vol 301 (1) ◽  
pp. C21-C30 ◽  
Author(s):  
Maoyun Sun ◽  
Xinhua Yan ◽  
Yun Bian ◽  
Anthony O. Caggiano ◽  
James P. Morgan

Identification of factors that direct embryonic stem (ES) cell (ESC) differentiation into functional cardiomyocytes is essential for successful use of ESC-based therapy for cardiac repair. Neuregulin-1 (NRG1) and microRNA play important roles in the cardiac differentiation of ESCs. Understanding how NRG1 regulates microRNA will provide new mechanistic insights into the role of NRG1 on ESCs. It may also lead to the discovery of novel microRNAs that are important for ESC cardiac differentiation. The objective of this study was to assess the microRNA expression profile during NRG1-induced ESC cardiac differentiation. Murine ESCs were incubated with a recombinant NRG1β or an inhibitor of ErbB2 or ErbB4 during hanging drop-induced cardiac differentiation. The expression of cardiac-specific markers and microRNAs was analyzed by RT-PCR and microRNA array, respectively. We found that the expression of NRG1 and the ErbB receptors was increased during hanging drop-induced cardiac differentiation of ESCs. NRG1 stimulation during a specific developmental window enhanced, while inhibition of the ErbB2 or ErbB4 receptor inhibited, cardiac differentiation of ESCs. NRG1 increased the expression of mmu-miR-296–3p and mmu-miR-200c*, and decreased mmu-miR-465b-5p. Inhibition of mmu-miR-296–3p or mmu-miR-200c* decreased, while inhibition of mmu-miR-465–5p increased, the differentiation of ESCs into the cardiac lineage. This is the first report demonstrating that microRNAs are differentially regulated by NRG1-ErbB signaling during cardiac differentiation of ESCs. This study has also identified new microRNAs that are important for ESC cardiac differentiation.


Sign in / Sign up

Export Citation Format

Share Document